1
|
Zhang S, Meor Azlan NF, Josiah SS, Zhou J, Zhou X, Jie L, Zhang Y, Dai C, Liang D, Li P, Li Z, Wang Z, Wang Y, Ding K, Wang Y, Zhang J. The role of SLC12A family of cation-chloride cotransporters and drug discovery methodologies. J Pharm Anal 2023; 13:1471-1495. [PMID: 38223443 PMCID: PMC10785268 DOI: 10.1016/j.jpha.2023.09.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Revised: 06/20/2023] [Accepted: 09/05/2023] [Indexed: 09/12/2023] Open
Abstract
The solute carrier family 12 (SLC12) of cation-chloride cotransporters (CCCs) comprises potassium chloride cotransporters (KCCs, e.g. KCC1, KCC2, KCC3, and KCC4)-mediated Cl- extrusion, and sodium potassium chloride cotransporters (N[K]CCs, NKCC1, NKCC2, and NCC)-mediated Cl- loading. The CCCs play vital roles in cell volume regulation and ion homeostasis. Gain-of-function or loss-of-function of these ion transporters can cause diseases in many tissues. In recent years, there have been considerable advances in our understanding of CCCs' control mechanisms in cell volume regulations, with many techniques developed in studying the functions and activities of CCCs. Classic approaches to directly measure CCC activity involve assays that measure the transport of potassium substitutes through the CCCs. These techniques include the ammonium pulse technique, radioactive or nonradioactive rubidium ion uptake-assay, and thallium ion-uptake assay. CCCs' activity can also be indirectly observed by measuring γ-aminobutyric acid (GABA) activity with patch-clamp electrophysiology and intracellular chloride concentration with sensitive microelectrodes, radiotracer 36Cl-, and fluorescent dyes. Other techniques include directly looking at kinase regulatory sites phosphorylation, flame photometry, 22Na+ uptake assay, structural biology, molecular modeling, and high-throughput drug screening. This review summarizes the role of CCCs in genetic disorders and cell volume regulation, current methods applied in studying CCCs biology, and compounds developed that directly or indirectly target the CCCs for disease treatments.
Collapse
Affiliation(s)
- Shiyao Zhang
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 363001, China
| | - Nur Farah Meor Azlan
- Institute of Biomedical and Clinical Sciences, Medical School, Faculty of Health and Life Sciences, University of Exeter, Exeter, EX4 4PS, UK
| | - Sunday Solomon Josiah
- Institute of Biomedical and Clinical Sciences, Medical School, Faculty of Health and Life Sciences, University of Exeter, Exeter, EX4 4PS, UK
| | - Jing Zhou
- Department of Neurology, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institute of Biological Science, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Xiaoxia Zhou
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 363001, China
| | - Lingjun Jie
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 363001, China
| | - Yanhui Zhang
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 363001, China
| | - Cuilian Dai
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 363001, China
| | - Dong Liang
- Aurora Discovery Inc., Foshan, Guangdong, 528300, China
| | - Peifeng Li
- Institute for Translational Medicine, Qingdao University, Qingdao, Shandong, 266021, China
| | - Zhengqiu Li
- School of Pharmacy, Jinan University, Guangzhou, 510632, China
| | - Zhen Wang
- State Key Laboratory of Chemical Biology, Research Center of Chemical Kinomics, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, 200032, China
| | - Yun Wang
- Department of Neurology, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institute of Biological Science, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Ke Ding
- State Key Laboratory of Chemical Biology, Research Center of Chemical Kinomics, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, 200032, China
| | - Yan Wang
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 363001, China
| | - Jinwei Zhang
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 363001, China
- Institute of Biomedical and Clinical Sciences, Medical School, Faculty of Health and Life Sciences, University of Exeter, Exeter, EX4 4PS, UK
- State Key Laboratory of Chemical Biology, Research Center of Chemical Kinomics, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, 200032, China
| |
Collapse
|
2
|
Bazzone A, Barthmes M, George C, Brinkwirth N, Zerlotti R, Prinz V, Cole K, Friis S, Dickson A, Rice S, Lim J, Fern Toh M, Mohammadi M, Pau D, Stone DJ, Renger JJ, Fertig N. A Comparative Study on the Lysosomal Cation Channel TMEM175 Using Automated Whole-Cell Patch-Clamp, Lysosomal Patch-Clamp, and Solid Supported Membrane-Based Electrophysiology: Functional Characterization and High-Throughput Screening Assay Development. Int J Mol Sci 2023; 24:12788. [PMID: 37628970 PMCID: PMC10454728 DOI: 10.3390/ijms241612788] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 08/09/2023] [Accepted: 08/12/2023] [Indexed: 08/27/2023] Open
Abstract
The lysosomal cation channel TMEM175 is a Parkinson's disease-related protein and a promising drug target. Unlike whole-cell automated patch-clamp (APC), lysosomal patch-clamp (LPC) facilitates physiological conditions, but is not yet suitable for high-throughput screening (HTS) applications. Here, we apply solid supported membrane-based electrophysiology (SSME), which enables both direct access to lysosomes and high-throughput electrophysiological recordings. In SSME, ion translocation mediated by TMEM175 is stimulated using a concentration gradient at a resting potential of 0 mV. The concentration-dependent K+ response exhibited an I/c curve with two distinct slopes, indicating the existence of two conducting states. We measured H+ fluxes with a permeability ratio of PH/PK = 48,500, which matches literature findings from patch-clamp studies, validating the SSME approach. Additionally, TMEM175 displayed a high pH dependence. Decreasing cytosolic pH inhibited both K+ and H+ conductivity of TMEM175. Conversely, lysosomal pH and pH gradients did not have major effects on TMEM175. Finally, we developed HTS assays for drug screening and evaluated tool compounds (4-AP, Zn as inhibitors; DCPIB, arachidonic acid, SC-79 as enhancers) using SSME and APC. Additionally, we recorded EC50 data for eight blinded TMEM175 enhancers and compared the results across all three assay technologies, including LPC, discussing their advantages and disadvantages.
Collapse
Affiliation(s)
- Andre Bazzone
- Nanion Technologies, Ganghoferstr. 70a, 80339 Munich, Germany (V.P.); (S.F.)
| | - Maria Barthmes
- Nanion Technologies, Ganghoferstr. 70a, 80339 Munich, Germany (V.P.); (S.F.)
| | - Cecilia George
- Nanion Technologies, Ganghoferstr. 70a, 80339 Munich, Germany (V.P.); (S.F.)
| | - Nina Brinkwirth
- Nanion Technologies, Ganghoferstr. 70a, 80339 Munich, Germany (V.P.); (S.F.)
| | - Rocco Zerlotti
- Nanion Technologies, Ganghoferstr. 70a, 80339 Munich, Germany (V.P.); (S.F.)
- RIGeL-Regensburg International Graduate School of Life Sciences, University of Regensburg, 93053 Regensburg, Germany
| | - Valentin Prinz
- Nanion Technologies, Ganghoferstr. 70a, 80339 Munich, Germany (V.P.); (S.F.)
| | - Kim Cole
- Nanion Technologies, Ganghoferstr. 70a, 80339 Munich, Germany (V.P.); (S.F.)
| | - Søren Friis
- Nanion Technologies, Ganghoferstr. 70a, 80339 Munich, Germany (V.P.); (S.F.)
| | - Alexander Dickson
- SB Drug Discovery, West of Scotland Science Park, Glasgow G20 0XA, UK; (A.D.); (S.R.)
| | - Simon Rice
- SB Drug Discovery, West of Scotland Science Park, Glasgow G20 0XA, UK; (A.D.); (S.R.)
| | - Jongwon Lim
- Cerevel Therapeutics, 222 Jacobs St, Cambridge, MA 02141, USA; (J.L.); (M.F.T.); (D.J.S.); (J.J.R.)
| | - May Fern Toh
- Cerevel Therapeutics, 222 Jacobs St, Cambridge, MA 02141, USA; (J.L.); (M.F.T.); (D.J.S.); (J.J.R.)
| | | | - Davide Pau
- SB Drug Discovery, West of Scotland Science Park, Glasgow G20 0XA, UK; (A.D.); (S.R.)
| | - David J. Stone
- Cerevel Therapeutics, 222 Jacobs St, Cambridge, MA 02141, USA; (J.L.); (M.F.T.); (D.J.S.); (J.J.R.)
| | - John J. Renger
- Cerevel Therapeutics, 222 Jacobs St, Cambridge, MA 02141, USA; (J.L.); (M.F.T.); (D.J.S.); (J.J.R.)
| | - Niels Fertig
- Nanion Technologies, Ganghoferstr. 70a, 80339 Munich, Germany (V.P.); (S.F.)
| |
Collapse
|
3
|
Putt KS, Du Y, Fu H, Zhang ZY. High-throughput screening strategies for space-based radiation countermeasure discovery. LIFE SCIENCES IN SPACE RESEARCH 2022; 35:88-104. [PMID: 36336374 DOI: 10.1016/j.lssr.2022.07.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 06/13/2022] [Accepted: 07/19/2022] [Indexed: 06/16/2023]
Abstract
As humanity begins to venture further into space, approaches to better protect astronauts from the hazards found in space need to be developed. One particular hazard of concern is the complex radiation that is ever present in deep space. Currently, it is unlikely enough spacecraft shielding could be launched that would provide adequate protection to astronauts during long-duration missions such as a journey to Mars and back. In an effort to identify other means of protection, prophylactic radioprotective drugs have been proposed as a potential means to reduce the biological damage caused by this radiation. Unfortunately, few radioprotectors have been approved by the FDA for usage and for those that have been developed, they protect normal cells/tissues from acute, high levels of radiation exposure such as that from oncology radiation treatments. To date, essentially no radioprotectors have been developed that specifically counteract the effects of chronic low-dose rate space radiation. This review highlights how high-throughput screening (HTS) methodologies could be implemented to identify such a radioprotective agent. Several potential target, pathway, and phenotypic assays are discussed along with potential challenges towards screening for radioprotectors. Utilizing HTS strategies such as the ones proposed here have the potential to identify new chemical scaffolds that can be developed into efficacious radioprotectors that are specifically designed to protect astronauts during deep space journeys. The overarching goal of this review is to elicit broader interest in applying drug discovery techniques, specifically HTS towards the identification of radiation countermeasures designed to be efficacious towards the biological insults likely to be encountered by astronauts on long duration voyages.
Collapse
Affiliation(s)
- Karson S Putt
- Institute for Drug Discovery, Purdue University, West Lafayette IN 47907 USA
| | - Yuhong Du
- Department of Pharmacology and Chemical Biology and Emory Chemical Biology Discovery Center, Emory University School of Medicine, Atlanta, GA 30322 USA
| | - Haian Fu
- Department of Pharmacology and Chemical Biology and Emory Chemical Biology Discovery Center, Emory University School of Medicine, Atlanta, GA 30322 USA
| | - Zhong-Yin Zhang
- Institute for Drug Discovery, Purdue University, West Lafayette IN 47907 USA; Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette IN 47907 USA.
| |
Collapse
|
4
|
High-Throughput Fluorescence Assays for Ion Channels and GPCRs. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1131:27-72. [DOI: 10.1007/978-3-030-12457-1_3] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
5
|
Crea F, De Stefano C, Foti C, Lando G, Milea D, Sammartano S. Alkali Metal Ion Complexes with Phosphates, Nucleotides, Amino Acids, and Related Ligands of Biological Relevance. Their Properties in Solution. Met Ions Life Sci 2016; 16:133-66. [PMID: 26860301 DOI: 10.1007/978-3-319-21756-7_5] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Alkali metal ions play very important roles in all biological systems, some of them are essential for life. Their concentration depends on several physiological factors and is very variable. For example, sodium concentrations in human fluids vary from quite low (e.g., 8.2 mmol dm(-3) in mature maternal milk) to high values (0.14 mol dm(-3) in blood plasma). While many data on the concentration of Na(+) and K(+) in various fluids are available, the information on other alkali metal cations is scarce. Since many vital functions depend on the network of interactions occurring in various biofluids, this chapter reviews their complex formation with phosphates, nucleotides, amino acids, and related ligands of biological relevance. Literature data on this topic are quite rare if compared to other cations. Generally, the stability of alkali metal ion complexes of organic and inorganic ligands is rather low (usually log K < 2) and depends on the charge of the ligand, owing to the ionic nature of the interactions. At the same time, the size of the cation is an important factor that influences the stability: very often, but not always (e.g., for sulfate), it follows the trend Li(+) > Na(+) > K(+) > Rb(+) > Cs(+). For example, for citrate it is: log K ML = 0.88, 0.80, 0.48, 0.38, and 0.13 at 25 °C and infinite dilution. Some considerations are made on the main aspects related to the difficulties in the determination of weak complexes. The importance of the alkali metal ion complexes was also studied in the light of modelling natural fluids and in the use of these cations as probes for different processes. Some empirical relationships are proposed for the dependence of the stability constants of Na(+) complexes on the ligand charge, as well as for correlations among log K values of NaL, KL or LiL species (L = generic ligand).
Collapse
Affiliation(s)
- Francesco Crea
- Dipartimento di Scienze Chimiche, Università di Messina, Viale F. Stagno d'Alcontres, 31, I-98166, Messina, Italy
| | - Concetta De Stefano
- Dipartimento di Scienze Chimiche, Università di Messina, Viale F. Stagno d'Alcontres, 31, I-98166, Messina, Italy
| | - Claudia Foti
- Dipartimento di Scienze Chimiche, Università di Messina, Viale F. Stagno d'Alcontres, 31, I-98166, Messina, Italy
| | - Gabriele Lando
- Dipartimento di Scienze Chimiche, Università di Messina, Viale F. Stagno d'Alcontres, 31, I-98166, Messina, Italy
| | - Demetrio Milea
- Dipartimento di Scienze Chimiche, Università di Messina, Viale F. Stagno d'Alcontres, 31, I-98166, Messina, Italy
| | - Silvio Sammartano
- Dipartimento di Scienze Chimiche, Università di Messina, Viale F. Stagno d'Alcontres, 31, I-98166, Messina, Italy.
| |
Collapse
|
6
|
High throughput screening technologies for ion channels. Acta Pharmacol Sin 2016; 37:34-43. [PMID: 26657056 DOI: 10.1038/aps.2015.108] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Accepted: 10/12/2015] [Indexed: 11/08/2022] Open
Abstract
Ion channels are involved in a variety of fundamental physiological processes, and their malfunction causes numerous human diseases. Therefore, ion channels represent a class of attractive drug targets and a class of important off-targets for in vitro pharmacological profiling. In the past decades, the rapid progress in developing functional assays and instrumentation has enabled high throughput screening (HTS) campaigns on an expanding list of channel types. Chronologically, HTS methods for ion channels include the ligand binding assay, flux-based assay, fluorescence-based assay, and automated electrophysiological assay. In this review we summarize the current HTS technologies for different ion channel classes and their applications.
Collapse
|
7
|
Farre C, George M, Brüggemann A, Fertig N. Ion channel screening - automated patch clamp on the rise. DRUG DISCOVERY TODAY. TECHNOLOGIES 2013; 5:e1-e34. [PMID: 24125503 DOI: 10.1016/j.ddtec.2008.12.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
8
|
Dunlop J, Peri R, Terstappen GC, Bowlby M. Functional screening of α7 nicotinic receptor ligands. Expert Opin Drug Discov 2013; 3:623-8. [PMID: 23506144 DOI: 10.1517/17460441.3.6.623] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND The α7 nicotinic acetylcholine receptor, a ligand-gated ion channel, is an attractive drug discovery target in schizophrenia and Alzheimer's disease. OBJECTIVE We have evaluated the various approaches to discovering ligands targeting the α7 nicotinic receptor to define the current paradigm driving drug discovery efforts in this area. METHODS Assays using functional read-outs as a consequence of α7 nicotinic receptor activation have been reviewed. CONCLUSION Functional assays using fluorescence-based optical methods in combination with direct electrophysiological recordings of channel function currently provide an integrated approach to the discovery of α7 nicotinic receptor targeted ligands.
Collapse
Affiliation(s)
- John Dunlop
- Neuroscience Discovery Research, Wyeth Research, CN-8000, Princeton, NJ 08543, USA +1 732 274 4193 ; +1 732 274 4755 ;
| | | | | | | |
Collapse
|
9
|
Shavkunov A, Panova N, Prasai A, Veselenak R, Bourne N, Stoilova-McPhie S, Laezza F. Bioluminescence methodology for the detection of protein-protein interactions within the voltage-gated sodium channel macromolecular complex. Assay Drug Dev Technol 2012; 10:148-60. [PMID: 22364545 DOI: 10.1089/adt.2011.413] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Protein-protein interactions are critical molecular determinants of ion channel function and emerging targets for pharmacological interventions. Yet, current methodologies for the rapid detection of ion channel macromolecular complexes are still lacking. In this study we have adapted a split-luciferase complementation assay (LCA) for detecting the assembly of the voltage-gated Na+ (Nav) channel C-tail and the intracellular fibroblast growth factor 14 (FGF14), a functionally relevant component of the Nav channelosome that controls gating and targeting of Nav channels through direct interaction with the channel C-tail. In the LCA, two complementary N-terminus and C-terminus fragments of the firefly luciferase were fused, respectively, to a chimera of the CD4 transmembrane segment and the C-tail of Nav1.6 channel (CD4-Nav1.6-NLuc) or FGF14 (CLuc-FGF14). Co-expression of CLuc-FGF14 and CD4-Nav1.6-NLuc in live cells led to a robust assembly of the FGF14:Nav1.6 C-tail complex, which was attenuated by introducing single-point mutations at the predicted FGF14:Nav channel interface. To evaluate the dynamic regulation of the FGF14:Nav1.6 C-tail complex by signaling pathways, we investigated the effect of kinase inhibitors on the complex formation. Through a platform of counter screenings, we show that the p38/MAPK inhibitor, PD169316, and the IκB kinase inhibitor, BAY 11-7082, reduce the FGF14:Nav1.6 C-tail complementation, highlighting a potential role of the p38MAPK and the IκB/NFκB pathways in controlling neuronal excitability through protein-protein interactions. We envision the methodology presented here as a new valuable tool to allow functional evaluations of protein-channel complexes toward probe development and drug discovery targeting ion channels implicated in human disorders.
Collapse
Affiliation(s)
- Alexander Shavkunov
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas 77555, USA
| | | | | | | | | | | | | |
Collapse
|
10
|
Biton B, Sethuramanujam S, Picchione KE, Bhattacharjee A, Khessibi N, Chesney F, Lanneau C, Curet O, Avenet P. The antipsychotic drug loxapine is an opener of the sodium-activated potassium channel slack (Slo2.2). J Pharmacol Exp Ther 2011; 340:706-15. [PMID: 22171093 DOI: 10.1124/jpet.111.184622] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Sodium-activated potassium (K(Na)) channels have been suggested to set the resting potential, to modulate slow after-hyperpolarizations, and to control bursting behavior or spike frequency adaptation (Trends Neurosci 28:422-428, 2005). One of the genes that encodes K(Na) channels is called Slack (Kcnt1, Slo2.2). Studies found that Slack channels were highly expressed in nociceptive dorsal root ganglion neurons and modulated their firing frequency (J Neurosci 30:14165-14172, 2010). Therefore, Slack channel openers are of significant interest as putative analgesic drugs. We screened the library of pharmacologically active compounds with recombinant human Slack channels expressed in Chinese hamster ovary cells, by using rubidium efflux measurements with atomic absorption spectrometry. Riluzole at 500 μM was used as a reference agonist. The antipsychotic drug loxapine and the anthelmintic drug niclosamide were both found to activate Slack channels, which was confirmed by using manual patch-clamp analyses (EC(50) = 4.4 μM and EC(50) = 2.9 μM, respectively). Psychotropic drugs structurally related to loxapine were also evaluated in patch-clamp experiments, but none was found to be as active as loxapine. Loxapine properties were confirmed at the single-channel level with recombinant rat Slack channels. In dorsal root ganglion neurons, loxapine was found to behave as an opener of native K(Na) channels and to increase the rheobase of action potential. This study identifies new K(Na) channel pharmacological tools, which will be useful for further Slack channel investigations.
Collapse
Affiliation(s)
- B Biton
- Exploratory Unit, Sanofi, 1 Avenue P Brossolette, 91385 Chilly-Mazarin Cedex, France.
| | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Su X, Young EWK, Underkofler HAS, Kamp TJ, January CT, Beebe DJ. Microfluidic cell culture and its application in high-throughput drug screening: cardiotoxicity assay for hERG channels. ACTA ACUST UNITED AC 2010; 16:101-11. [PMID: 21131594 DOI: 10.1177/1087057110386218] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Evaluation of drug cardiotoxicity is essential to the safe development of novel pharmaceuticals. Assessing a compound's risk for prolongation of the surface electrocardiographic QT interval and hence risk for life-threatening arrhythmias is mandated before approval of nearly all new pharmaceuticals. QT prolongation has most commonly been associated with loss of current through hERG (human ether-a-go-go related gene) potassium ion channels due to direct block of the ion channel by drugs or occasionally by inhibition of the plasma membrane expression of the channel protein. To develop an efficient, reliable, and cost-effective hERG screening assay for detecting drug-mediated disruption of hERG membrane trafficking, the authors demonstrate the use of microfluidic-based systems to improve throughput and lower cost of current methods. They validate their microfluidics array platform in polystyrene (PS), cyclo-olefin polymer (COP), and polydimethylsiloxane (PDMS) microchannels for drug-induced disruption of hERG trafficking by culturing stably transfected HEK cells that overexpressed hERG (WT-hERG) and studying their morphology, proliferation rates, hERG protein expression, and response to drug treatment. Results show that WT-hERG cells readily proliferate in PS, COP, and PDMS microfluidic channels. The authors demonstrated that conventional Western blot analysis was possible using cell lysate extracted from a single microchannel. The Western blot analysis also provided important evidence that WT-hERG cells cultured in microchannels maintained regular (well plate-based) expression of hERG. The authors further show that experimental procedures can be streamlined by using direct in-channel immunofluorescence staining in conjunction with detection using an infrared scanner. Finally, treatment of WT-hERG cells with 5 different drugs suggests that PS (and COP) microchannels were more suitable than PDMS microchannels for drug screening applications, particularly for tests involving hydrophobic drug molecules.
Collapse
Affiliation(s)
- Xiaojing Su
- Department of Biomedical Engineering and Wisconsin Institutes for Medical Research, University of Wisconsin-Madison, Madison, WI, USA
| | | | | | | | | | | |
Collapse
|
12
|
Meyer T, Stuerz K, Guenther E, Edamura M, Kraushaar U. Cardiac slices as a predictive tool for arrhythmogenic potential of drugs and chemicals. Expert Opin Drug Metab Toxicol 2010; 6:1461-75. [PMID: 21067457 DOI: 10.1517/17425255.2010.526601] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
IMPORTANCE OF THE FIELD cardiac arrhythmia represents one of the primary safety pharmacological concerns in drug development. The most prominent example is drug induced ventricular tachycardia of the Torsade des Pointes type. The mechanism how this type of arrhythmia develops is a complex multi-cellular phenomenon. It can only be insufficiently reflected by cellular or molecular assays. However, organ models - such as Langendorff hearts - or in vivo experiments are expensive and time consuming and not suitable for assays requiring an increased throughput. AREAS COVERED IN THIS REVIEW here, we describe and review an assay bridging the gap between cardiomyocyte based assays and organ based systems - cardiac slices. This assay is reviewed in direct comparison with established safety pharmacological assays. WHAT THE READER WILL GAIN while slices have played an important role in brain research for > 2 decades, cardiac slices are experiencing a renaissance due to the novel challenges in safety pharmacology just in the last few years. Cardiac slices can be cultured and recorded over several days. It is possible to access electrophysiological data with a high number of electrodes - up to 256 electrodes - embedded in the surface of a microelectrode array. TAKE HOME MESSAGE cardiac slices close the gap between cellular and organ based assays in cardiac safety pharmacology. The tissue properties of a functional cardiac syncytium are more accurately reflected by a slice rather than a single cell.
Collapse
Affiliation(s)
- Thomas Meyer
- Multi Channel Systems MCS GmbH, Aspenhaustr. 21, 72770 Reutlingen, Germany.
| | | | | | | | | |
Collapse
|
13
|
Abstract
For every movement, heartbeat and thought, ion channels need to open and close. It is therefore not surprising that their malfunctioning leads to serious diseases. Currently, only approximately 10% of drugs, with a market value in excess of US$10 billion, act on ion channels. The systematic exploitation of this target class has started, enabled by novel assay technologies and fundamental advances of the structural and mechanistic understanding of channel function. The latter, which was rewarded with the Nobel Prize in 2003, has opened up an avenue for rational drug design. In this review we provide an overview of the current repertoire of screening technologies that has evolved to drive ion channel-targeted drug discovery towards new medicines of the future.
Collapse
|
14
|
Vasilyev DV, Shan QJ, Lee YT, Soloveva V, Nawoschik SP, Kaftan EJ, Dunlop J, Mayer SC, Bowlby MR. A Novel High-Throughput Screening Assay for HCN Channel Blocker Using Membrane Potential-Sensitive Dye and FLIPR. ACTA ACUST UNITED AC 2009; 14:1119-28. [DOI: 10.1177/1087057109345526] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Hyperpolarization-activated cation nonselective (HCN) channels represent an interesting group of targets for drug development. In this study, the authors report the development of a novel membrane potential-sensitive dye (MPSD) assay for HCN channel modulators that has been miniaturized into 384-well fluorescent imaging plate reader (FLIPR) high-throughput screening (HTS) format. When optimized (by cell plating density, plate type, cell recovery from cryopreservation), the wellto-well signal variability was low, with a Z' = 0.73 and coefficient of variation = 6.4%, whereas the MPSD fluorescence signal amplitude was -23,700 ± 1500 FLIPR3 relative fluorescence units (a linear relationship was found between HCN1 MPSD fluorescence signal and the cell plating density) and was completely blocked by 30 µM ZD7288. The assay tolerated up to 1% DMSO, inclusion of which did not significantly change the signal kinetics or amplitude. A single-concentration screening of an ion channel-focused library composed of 4855 compounds resulted in 89 HCN1 blocker hits, 51 of which were subsequently analyzed with an 8-point concentration-response analysis on the IonWorks HT electrophysiology platform. The correlation between MPSD and the electrophysiology assay was moderate, as shown by the linear regression analysis (r2 = 0.56) between the respective IC50s obtained using these 2 assays. The reported HTS-compatible HCN channel blocker assay can serve as a tool in drug discovery in the pursuit of HCN channel isoform-selective small molecules that could be used in the development of clinically relevant compounds. (Journal of Biomolecular Screening 2009:1119-1128)
Collapse
Affiliation(s)
| | - Qin J. Shan
- 'Discovery Neuroscience, Wyeth Research, Princeton, New Jersey
| | - Yan T. Lee
- 'Discovery Neuroscience, Wyeth Research, Princeton, New Jersey
| | - Veronica Soloveva
- Chemical and Screening Sciences, Wyeth Research, Princeton, New Jersey
| | | | | | - John Dunlop
- 'Discovery Neuroscience, Wyeth Research, Princeton, New Jersey
| | - Scott C. Mayer
- Chemical and Screening Sciences, Wyeth Research, Princeton, New Jersey
| | - Mark R. Bowlby
- 'Discovery Neuroscience, Wyeth Research, Princeton, New Jersey
| |
Collapse
|
15
|
Abstract
K(+) channels are revered for their universal action of suppressing electrical activity in nerve and muscle, as well as regulating salt and water transport in epithelial tissues involved in metabolism and digestion. These multisubunit membrane-embedded proteins carry out their physiological chore, selectively allowing the passage of potassium across the membrane, in response to changes in membrane voltage and ligand concentration. Elucidating the diverse gating properties of K(+) channels is of great biological interest since their molecular motions provide insight into how these structurally similar proteins function in a wide variety of tissues. Armed with patch clamps, chart recorders, and now high-resolution structures, electrophysiologists have been dipping into the top tray of the chemist's tool box: synthesizing cysteine-modifying agents and organic cations and grinding up insects, spiders, and other vermin to isolate natural products to poke, probe, and prod K(+) channels. Recently, there has been further cross-fertilization between chemists and K(+) channelologists, resulting in greater accessibility to more elaborate synthetic methodologies and screening approaches. In this review, we catalogue the evolution of chemical tools and approaches that have been utilized to elucidate the mechanistic underpinnings of K(+) channel biology.
Collapse
Affiliation(s)
- Christopher A Ahern
- Department of Anesthesiology, University of British Columbia, 2350 Health Science Mall, Vancouver, BC V6T 1Z3, Canada.
| | | |
Collapse
|
16
|
Trivedi S, Dekermendjian K, Julien R, Huang J, Lund PE, Krupp J, Kronqvist R, Larsson O, Bostwick R. Cellular HTS assays for pharmacological characterization of Na(V)1.7 modulators. Assay Drug Dev Technol 2008; 6:167-79. [PMID: 18078380 DOI: 10.1089/adt.2007.090] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Ion channels are challenging targets in the early phases of the drug discovery process, especially because of the lack of technologies available to screen large numbers of compounds in functionally relevant assays. The electrophysiological patch-clamp technique, which is the gold standard for studying ion channels, has low throughput and is not amenable to screening large numbers of compounds. However, for random high-throughput screening (HTS) of compounds against ion channel targets, a number of functional cellular assays have become available during the last few years. Here we use the sodium channel NaV1.7 stably expressed in human embryonic kidney 293 cells and compare three HTS assays-a Li flux atomic absorption spectroscopy (AAS) assay, a fluorescent imaging plate reader (FLIP, Molecular Devices, Sunnyvale, CA) membrane potential assay, and a fluorescence resonance energy transfer (FRET)-based membrane potential assay-to an automated electrophysiological assay (the Ionworks HT [Molecular Devices] platform) and characterize 11 known NaV inhibitors. Our results show that all three HTS assays are suitable for identification of NaV1.7 inhibitors, but as an HTS assay the Li-AAS assay is more robust with higher Z' values than the FLIPR and FRET-based membrane potential assays. Furthermore, there was a better correlation between the Ionworks assay and the Li-AAS assay regarding the potency of the NaV inhibitors investigated. This paper describes the first comparison between all the HTS assays available today to study voltage-gated NaVs, and the results suggest that the Li-AAS assay is more suited as a first HTS assay when starting an NaV drug discovery campaign.
Collapse
Affiliation(s)
- Shephali Trivedi
- HTS Center and Global Support Department, AstraZeneca Pharmaceuticals, Wilmington, DE 19850, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Dunlop J, Bowlby M, Peri R, Vasilyev D, Arias R. High-throughput electrophysiology: an emerging paradigm for ion-channel screening and physiology. Nat Rev Drug Discov 2008; 7:358-68. [PMID: 18356919 DOI: 10.1038/nrd2552] [Citation(s) in RCA: 341] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Ion channels represent highly attractive targets for drug discovery and are implicated in a diverse range of disorders, in particular in the central nervous and cardiovascular systems. Moreover, assessment of cardiac ion-channel activity of new chemical entities is now an integral component of drug discovery programmes to assess potential for cardiovascular side effects. Despite their attractiveness as drug discovery targets ion channels remain an under-exploited target class, which is in large part due to the labour-intensive and low-throughput nature of patch-clamp electrophysiology. This Review provides an update on the current state-of-the-art for the various automated electrophysiology platforms that are now available and critically evaluates their impact in terms of ion-channel screening, lead optimization and the assessment of cardiac ion-channel safety liability.
Collapse
Affiliation(s)
- John Dunlop
- Neuroscience Discovery Research, Wyeth Research, CN-8000, Princeton, New Jersey 08543, USA.
| | | | | | | | | |
Collapse
|
18
|
Inglese J, Johnson RL, Simeonov A, Xia M, Zheng W, Austin CP, Auld DS. High-throughput screening assays for the identification of chemical probes. Nat Chem Biol 2007; 3:466-79. [PMID: 17637779 DOI: 10.1038/nchembio.2007.17] [Citation(s) in RCA: 434] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
High-throughput screening (HTS) assays enable the testing of large numbers of chemical substances for activity in diverse areas of biology. The biological responses measured in HTS assays span isolated biochemical systems containing purified receptors or enzymes to signal transduction pathways and complex networks functioning in cellular environments. This Review addresses factors that need to be considered when implementing assays for HTS and is aimed particularly at investigators new to this field. We discuss assay design strategies, the major detection technologies and examples of HTS assays for common target classes, cellular pathways and simple cellular phenotypes. We conclude with special considerations for configuring sensitive, robust, informative and economically feasible HTS assays.
Collapse
MESH Headings
- Animals
- Catalysis
- Chemistry, Pharmaceutical/instrumentation
- Chemistry, Pharmaceutical/methods
- Drug Design
- Drug Evaluation, Preclinical/instrumentation
- Drug Evaluation, Preclinical/methods
- Enzymes/chemistry
- Humans
- Ions
- Kinetics
- Models, Biological
- Models, Chemical
- Receptors, G-Protein-Coupled/metabolism
- Signal Transduction
- Technology, Pharmaceutical/instrumentation
- Technology, Pharmaceutical/methods
- Transcription, Genetic
Collapse
Affiliation(s)
- James Inglese
- US National Institutes of Health Chemical Genomics Center, National Institutes of Health, 9800 Medical Center Drive, Bethesda, Maryland 20892-3370, USA.
| | | | | | | | | | | | | |
Collapse
|
19
|
Dunlop J, Roncarati R, Jow B, Bothmann H, Lock T, Kowal D, Bowlby M, Terstappen GC. In vitro screening strategies for nicotinic receptor ligands. Biochem Pharmacol 2007; 74:1172-81. [PMID: 17706607 DOI: 10.1016/j.bcp.2007.07.006] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2007] [Revised: 06/29/2007] [Accepted: 07/03/2007] [Indexed: 11/17/2022]
Abstract
A common historical strategy to the discovery of nicotinic receptor ligands has involved the use of radioligand-binding assays for ligand identification in combination with two-electrode voltage clamp in Xenopus oocytes for electrophysiological characterization. More recently, higher-throughput methodologies have replaced these approaches to accommodate screening of large compound libraries and to provide increased capacity for electrophysiological profiling in mammalian cell lines. We, and others, have implemented cell-based screening assays using the fluorometric imaging plate reader (FLIPR) for primary and lead optimization screening of nicotinic receptor agonists and positive allosteric modulators (PAMs). Using GH4C1 cells expressing the rat alpha7 nicotinic receptor, both acetylcholine and nicotine produced concentration-dependent elevations of intracellular calcium with EC(50) values of 5.5 and 1.6 microM, respectively. PAM activity was robustly detected using the FLIPR assay; for example, the known alpha7 receptor PAM 5-hydroxyindole failed to directly activate the receptor but produced a leftward shift of the nicotine concentration-response curve in combination with a potentiation of the maximum evoked response to nicotine. Electrophysiological confirmation of agonist activity was achieved using the Dynaflow rapid perfusion system and patch clamp in the same GH4C1 cell expression system. Estimated EC(50) values for acetylcholine-evoked currents in GH4C1/alpha7 cells were 55 and 576 microM for area-under-the-curve (AUC) and maximum peak height calculations, respectively. Similarly, PAM activity was confirmed using electrophysiological recordings while also allowing for the mechanistic discrimination of compounds, not possible using the FLIPR assay. Specifically, PAMs capable of slowing the rapid desensitization of alpha7 receptors to different extents were discernable in these studies. Further improvements in the capacity to screen compounds using electrophysiology has been achieved by implementation of high-throughput gigaohm quality recording systems such as the QPatch and PatchXpress where agonist EC(50) values are highly comparable to those obtained using conventional manual patch clamp.
Collapse
Affiliation(s)
- John Dunlop
- Neuroscience Discovery Research, Wyeth Research, CN-8000, Princeton, NJ 08543, USA.
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Vasilyev D, Merrill T, Iwanow A, Dunlop J, Bowlby M. A novel method for patch-clamp automation. Pflugers Arch 2006; 452:240-7. [PMID: 16596408 DOI: 10.1007/s00424-005-0029-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2005] [Revised: 10/19/2005] [Accepted: 11/10/2005] [Indexed: 11/28/2022]
Abstract
An increasing demand of the pharmaceutical industry for automated electrophysiological stations for ion channel drug discovery has recently resulted in the development of several commercial platforms for secondary and safety screening of ion channel modulators. These commercial systems have demonstrated an enhanced throughput, however, often at the expense of some quality-sensitive aspects of traditional patch-clamp recordings. To improve data quality and content, we have developed a patch-clamp robot that fully automates manual patch-clamp recordings, including patch pipette handling, gigaseal formation, obtaining whole-cell or perforated-cell configuration, drug application, and data acquisition. Utilization of glass micropipettes results in high-quality electrophysiological recordings with an overall success rate of about 30% in perforated-cell mode. A fast drug application system with low volume requirements (1-1.5 ml) allows the study of ligand-gated ion channels on a millisecond scale. As proof-of-concept, we present two assays developed for voltage-gated human ether-a-go-go-related and ligand-gated alpha(7) nicotinic receptor ion channels. The system throughput was a single concentration-response curve every 30-40 min or 12-17 6-point concentration-response curves daily, representing a significant improvement of typical manual patch-clamp throughput. This system represents an efficient method for patch-clamp automation without the need for a complex and expensive electrophysiological set-up for cell visualization.
Collapse
Affiliation(s)
- D Vasilyev
- Discovery Neuroscience, Wyeth Research, CN 8000, Princeton, NJ 08543-8000, USA.
| | | | | | | | | |
Collapse
|
21
|
Terstappen GC. Ion channel screening technologies today. DRUG DISCOVERY TODAY. TECHNOLOGIES 2005; 2:133-140. [PMID: 24981840 DOI: 10.1016/j.ddtec.2005.05.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
For every heartbeat, movement and thought, ion channels have to open and close, and thus, it is not surprising that malfunctioning of these membrane proteins leads to serious diseases. Today, only 7% of all marketed drugs act on ion channels but the systematic exploitation of this important target class has started mainly enabled by novel screening technologies. Thus, the discovery of selective and state-dependent drugs is on the horizon, hopefully leading to effective novel medicines.:
Collapse
Affiliation(s)
- Georg C Terstappen
- Sienabiotech S.p.A., Discovery Research, Via Fiorentina 1, 53100 Siena, Italy.
| |
Collapse
|